151
|
An aPPARent Functional Consequence in Skeletal Muscle Physiology via Peroxisome Proliferator-Activated Receptors. Int J Mol Sci 2018; 19:ijms19051425. [PMID: 29747466 PMCID: PMC5983589 DOI: 10.3390/ijms19051425] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/05/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle comprises 30–40% of the total body mass and plays a central role in energy homeostasis in the body. The deregulation of energy homeostasis is a common underlying characteristic of metabolic syndrome. Over the past decades, peroxisome proliferator-activated receptors (PPARs) have been shown to play critical regulatory roles in skeletal muscle. The three family members of PPAR have overlapping roles that contribute to the myriad of processes in skeletal muscle. This review aims to provide an overview of the functions of different PPAR members in energy homeostasis as well as during skeletal muscle metabolic disorders, with a particular focus on human and relevant mouse model studies.
Collapse
|
152
|
Lund MT, Larsen S, Hansen M, Courraud J, Floyd AK, Støckel M, Helge JW, Dela F. Mitochondrial respiratory capacity remains stable despite a comprehensive and sustained increase in insulin sensitivity in obese patients undergoing gastric bypass surgery. Acta Physiol (Oxf) 2018; 223:e13032. [PMID: 29330917 DOI: 10.1111/apha.13032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 12/15/2022]
Abstract
AIM It has been proposed, but not yet demonstrated by convincing evidence in published articles, that insulin resistance and mitochondrial respiratory function are causally related physiological phenomena. Here, we tested the prediction that weight loss-induced increase in insulin sensitivity will correlate with a corresponding change in mitochondrial respiratory capacity over the same time period. METHODS Insulin sensitivity was evaluated using the hyperinsulinaemic-euglycaemic clamp technique, and skeletal muscle mitochondrial respiratory capacity was evaluated by high-resolution respirometry in 26 patients with obesity. Each experiment was performed ~2 months and 1-2 weeks before, and ~4 and ~19 months after Roux-en-Y gastric bypass (RYGB) surgery. RESULTS A substantial weight loss was observed in all patients, and insulin sensitivity increased in all patients over the 21-months time period of the study. In contrast, skeletal muscle mitochondrial respiratory capacity, intrinsic mitochondrial respiratory capacity and mitochondrial content remained unchanged over the same time period. CONCLUSION Among obese patients with and without type 2 diabetes undergoing RYGB surgery, intrinsic mitochondrial respiratory capacity in skeletal muscle is not correlated with insulin sensitivity before or after the surgical intervention. Mitochondrial respiratory function may not be germane to the pathophysiology and/or aetiology of obesity and/or type 2 diabetes.
Collapse
Affiliation(s)
- M. T. Lund
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Surgery; Holbak Hospital; Holbak Denmark
| | - S. Larsen
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
| | - M. Hansen
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
| | - J. Courraud
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Danish Center for Newborn screening; Department of Congenital Disorders; Statens Serum Institut; Copenhagen Denmark
| | - A. K. Floyd
- Department of Surgery; Holbak Hospital; Holbak Denmark
| | - M. Støckel
- Department of Surgery; Herlev University Hospital; Herlev Denmark
| | - J. W. Helge
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
| | - F. Dela
- Xlab; Center for Healthy Aging; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Geriatrics; Bispebjerg University Hospital; Copenhagen Denmark
| |
Collapse
|
153
|
Sullivan EM, Pennington ER, Green WD, Beck MA, Brown DA, Shaikh SR. Mechanisms by Which Dietary Fatty Acids Regulate Mitochondrial Structure-Function in Health and Disease. Adv Nutr 2018; 9:247-262. [PMID: 29767698 PMCID: PMC5952932 DOI: 10.1093/advances/nmy007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/02/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are the energy-producing organelles within a cell. Furthermore, mitochondria have a role in maintaining cellular homeostasis and proper calcium concentrations, building critical components of hormones and other signaling molecules, and controlling apoptosis. Structurally, mitochondria are unique because they have 2 membranes that allow for compartmentalization. The composition and molecular organization of these membranes are crucial to the maintenance and function of mitochondria. In this review, we first present a general overview of mitochondrial membrane biochemistry and biophysics followed by the role of different dietary saturated and unsaturated fatty acids in modulating mitochondrial membrane structure-function. We focus extensively on long-chain n-3 (ω-3) polyunsaturated fatty acids and their underlying mechanisms of action. Finally, we discuss implications of understanding molecular mechanisms by which dietary n-3 fatty acids target mitochondrial structure-function in metabolic diseases such as obesity, cardiac-ischemia reperfusion injury, obesity, type 2 diabetes, nonalcoholic fatty liver disease, and select cancers.
Collapse
Affiliation(s)
- E Madison Sullivan
- Department of Biochemistry and Molecular Biology and
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Edward Ross Pennington
- Department of Biochemistry and Molecular Biology and
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
- Department of Nutrition, The University of North Carolina at Chapel Hill, Gillings School of Global Public Health and School of Medicine, Chapel Hill, NC
| | - William D Green
- Department of Nutrition, The University of North Carolina at Chapel Hill, Gillings School of Global Public Health and School of Medicine, Chapel Hill, NC
| | - Melinda A Beck
- Department of Nutrition, The University of North Carolina at Chapel Hill, Gillings School of Global Public Health and School of Medicine, Chapel Hill, NC
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech Corporate Research Center, Blacksburg, VA
| | - Saame Raza Shaikh
- Department of Nutrition, The University of North Carolina at Chapel Hill, Gillings School of Global Public Health and School of Medicine, Chapel Hill, NC
| |
Collapse
|
154
|
Gan L, Ma D, Li M, Yang FC, Rogers RS, Wheatley JL, Koch LG, Britton SL, Thyfault JP, Geiger PC, Stanford JA. Region-specific differences in bioenergetic proteins and protein response to acute high fat diet in brains of low and high capacity runner rats. Neurosci Lett 2018. [PMID: 29522838 DOI: 10.1016/j.neulet.2018.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Aerobic capacity is a strong predictor of mortality. Low capacity runner (LCR) rats exhibit reduced mitochondrial function in peripheral organs. A high fat diet (HFD) can worsen metabolic phenotype in LCR rats. Little is known about metabolic changes in the brains of these rats, however. This study examined protein markers of mitochondrial function and metabolism as a function of aerobic running capacity and an acute HFD in four brain regions: the striatum, hippocampus, hypothalamus, and substantia nigra. After 3 days HFD or chow diets, we measured peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1-α), nuclear respiratory factors 1 (Nrf-1), mitochondrial transcription factor A (TFAM), and phosphorylated (activated) AMP-activated protein kinase (p-AMPK) protein levels in the four brain regions. LCR rats exhibited lower levels of mitochondrial proteins (PGC1-α, Nrf-1, TFAM), and greater p-AMPK, in striatum, but not in the other brain regions. Mitochondrial protein levels were greater in HFD LCR striatum, while p-AMPK was lower in this group. Markers of lower mitochondrial biogenesis and increased metabolic demand were limited to the LCR striatum, which nevertheless maintained the capacity to respond to an acute HFD challenge.
Collapse
Affiliation(s)
- Li Gan
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Delin Ma
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Min Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Fu-Chen Yang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Robert S Rogers
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joshua L Wheatley
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Research Service, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Paige C Geiger
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John A Stanford
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
155
|
Potential Roles of n-3 PUFAs during Skeletal Muscle Growth and Regeneration. Nutrients 2018; 10:nu10030309. [PMID: 29510597 PMCID: PMC5872727 DOI: 10.3390/nu10030309] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 01/06/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), which are commonly found in fish oil supplements, are known to possess anti-inflammatory properties and more recently alter skeletal muscle function. In this review, we discuss novel findings related to how n-3 PUFAs modulate molecular signaling responsible for growth and hypertrophy as well as the activity of muscle stem cells. Muscle stem cells commonly known as satellite cells, are primarily responsible for driving the skeletal muscle repair process to potentially damaging stimuli, such as mechanical stress elicited by exercise contraction. To date, there is a paucity of human investigations related to the effects of n-3 PUFAs on satellite cell content and activity. Based on current in vitro investigations, this review focuses on novel mechanisms linking n-3 PUFA’s to satellite cell activity and how they may improve muscle repair. Understanding the role of n-3 PUFAs during muscle growth and regeneration in association with exercise could lead to the development of novel supplementation strategies that increase muscle mass and strength, therefore possibly reducing the burden of muscle wasting with age.
Collapse
|
156
|
Schönke M, Björnholm M, Chibalin AV, Zierath JR, Deshmukh AS. Proteomics Analysis of Skeletal Muscle from Leptin-Deficient ob/ob Mice Reveals Adaptive Remodeling of Metabolic Characteristics and Fiber Type Composition. Proteomics 2018; 18:e1700375. [PMID: 29350465 DOI: 10.1002/pmic.201700375] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/07/2018] [Indexed: 11/10/2022]
Abstract
Skeletal muscle insulin resistance, an early metabolic defect in the pathogenesis of type 2 diabetes (T2D), may be a cause or consequence of altered protein expression profiles. Proteomics technology offers enormous promise to investigate molecular mechanisms underlying pathologies, however, the analysis of skeletal muscle is challenging. Using state-of-the-art multienzyme digestion and filter-aided sample preparation (MED-FASP) and a mass spectrometry (MS)-based workflow, we performed a global proteomics analysis of skeletal muscle from leptin-deficient, obese, insulin resistant (ob/ob) and lean mice in mere two fractions in a short time (8 h per sample). We identified more than 6000 proteins with 118 proteins differentially regulated in obesity. This included protein kinases, phosphatases, and secreted and fiber type associated proteins. Enzymes involved in lipid metabolism in skeletal muscle from ob/ob mice were increased, providing evidence against reduced fatty acid oxidation in lipid-induced insulin resistance. Mitochondrial and peroxisomal proteins, as well as components of pyruvate and lactate metabolism, were increased. Finally, the skeletal muscle proteome from ob/ob mice displayed a shift toward the "slow fiber type." This detailed characterization of an obese rodent model of T2D demonstrates an efficient workflow for skeletal muscle proteomics, which may easily be adapted to other complex tissues.
Collapse
Affiliation(s)
- Milena Schönke
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
157
|
Toledo FGS, Johannsen DL, Covington JD, Bajpeyi S, Goodpaster B, Conley KE, Ravussin E. Impact of prolonged overfeeding on skeletal muscle mitochondria in healthy individuals. Diabetologia 2018; 61:466-475. [PMID: 29150696 PMCID: PMC5770194 DOI: 10.1007/s00125-017-4496-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/17/2017] [Indexed: 12/01/2022]
Abstract
AIMS/HYPOTHESES Reduced mitochondrial capacity in skeletal muscle has been observed in obesity and type 2 diabetes. In humans, the aetiology of this abnormality is not well understood but the possibility that it is secondary to the stress of nutrient overload has been suggested. To test this hypothesis, we examined whether sustained overfeeding decreases skeletal muscle mitochondrial content or impairs function. METHODS Twenty-six healthy volunteers (21 men, 5 women, age 25.3 ± 4.5 years, BMI 25.5 ± 2.4 kg/m2) underwent a supervised protocol consisting of 8 weeks of high-fat overfeeding (40% over baseline energy requirements). Before and after overfeeding, we measured systemic fuel oxidation by indirect calorimetry and performed skeletal muscle biopsies to measure mitochondrial gene expression, content and function in vitro. Mitochondrial function in vivo was measured by 31P NMR spectroscopy. RESULTS With overfeeding, volunteers gained 7.7 ± 1.8 kg (% change 9.8 ± 2.3). Overfeeding increased fasting NEFA, LDL-cholesterol and insulin concentrations. Indirect calorimetry showed a shift towards greater reliance on lipid oxidation. In skeletal muscle tissue, overfeeding increased ceramide content, lipid droplet content and perilipin-2 mRNA expression. Phosphorylation of AMP-activated protein kinase was decreased. Overfeeding increased mRNA expression of certain genes coding for mitochondrial proteins (CS, OGDH, CPT1B, UCP3, ANT1). Despite the stress of nutrient overload, mitochondrial content and mitochondrial respiration in muscle did not change after overfeeding. Similarly, overfeeding had no effect on either the emission of reactive oxygen species or on mitochondrial function in vivo. CONCLUSIONS/INTERPRETATION Skeletal muscle mitochondria are significantly resilient to nutrient overload. The lower skeletal muscle mitochondrial oxidative capacity in human obesity is likely to be caused by reasons other than nutrient overload per se. TRIAL REGISTRATION ClinicalTrials.gov NCT01672632.
Collapse
Affiliation(s)
- Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1054, Pittsburgh, PA, 15261, USA.
| | | | | | - Sudip Bajpeyi
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
- Department of Kinesiology, University of Texas El Paso, El Paso, TX, USA
| | - Bret Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Orlando, FL, USA
| | - Kevin E Conley
- University of Washington Medical Center, Seattle, WA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
158
|
Choi JW, Ohn JH, Jung HS, Park YJ, Jang HC, Chung SS, Park KS. Carnitine induces autophagy and restores high-fat diet-induced mitochondrial dysfunction. Metabolism 2018; 78:43-51. [PMID: 28966077 DOI: 10.1016/j.metabol.2017.09.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/12/2017] [Accepted: 09/23/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Autophagy is suppressed in skeletal muscle and the liver with insulin resistance induced by a high-fat diet. Autophagy is essential for maintaining mitochondrial function, and dysfunctional mitochondria are associated with insulin resistance. As carnitine treatment is well known to improve insulin resistance by promoting mitochondrial function, we investigated if carnitine affects autophagy in the skeletal muscle of a high-fat diet-induced rodent model of obesity. RESULTS After 6weeks on a high-fat diet (48kcal% fat), mice developed glucose intolerance, and the gastrocnemius muscle showed a decrease in insulin signaling and mitochondrial function, which was reversed after carnitine (100mg/kg/day) treatment by oral gavage for 2weeks. Swollen mitochondria with destroyed cristae were observed in the skeletal muscle of high-fat diet-fed mice but were not there after carnitine treatment. High-fat diet decreased LC3B-II, a marker of autophagosome formation, and increased sequestosome 1 (SQSTM1), expression of which was reversed after carnitine treatment. In C2C12 myotubes, prolonged treatment with palmitate suppressed autophagy, which was relieved by carnitine treatment. However, the induction of autophagy by carnitine in C2C12 myotubes was not observed after knock-down of peroxisome proliferator-activated receptor γ (PPARγ), which is known to regulate autophagy. CONCLUSION We conclude that the removal of dysfunctional mitochondria by induction of autophagy through PPARγ may be a novel mechanism by which carnitine improves insulin resistance and mitochondrial dysfunction in obesity.
Collapse
Affiliation(s)
- Jin Woo Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Hye Seung Jung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hak Chul Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Sung Soo Chung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Kyong Soo Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
159
|
Saleh N, Elayan HE, Zihlif M. THE EFFECT OF SALBUTAMOL ON PGC-1 α AND GLUT4 mRNA EXPRESSION IN THE LIVER AND MUSCLE OF ELDERLY DIABETIC MICE. ACTA ENDOCRINOLOGICA-BUCHAREST 2018; 14:184-191. [PMID: 31149256 DOI: 10.4183/aeb.2018.184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) plays an important role in the regulation of cellular energy metabolism, and it is involved in obesity and type 2 diabetes mellitus (T2DM). Its expression is elevated in the liver of T2DM mouse models. Literature reports show that chronic β2 stimulation improved insulin sensitivity in T2DM. Objectives We aimed to test the hypotheses that chronic β2 stimulation-induced improvement in insulin sensitivity involves changes in the expression of PGC-1α and glucose transporter 4 (GLUT4). Animals and Methods We fed a locally inbred, 8 months old mice, a high fat diet (HFD) to induce diabetes. These mice gained weight and became insulin resistant. The β2 agonist salbutamol had a beneficial effect on both glucose tolerance and insulin sensitivity after 4 weeks. Results Salbutamol beneficial effect persisted after 4 weeks of its discontinuation. HFD caused an up regulation of the hepatic PGC-1 α expression by 5.23 folds (P< 0.041) and salbutamol reversed this effect and caused a down regulation by 30.3 folds (P< 0.0001). PGC-1 α and GLUT4 expression in the muscle was not affected by salbutamol (P> 0.05). Conclusion Down regulation of the liver's PGC-1 α contributes to the beneficial effect of the chronic β2 stimulation on glucose tolerance and insulin sensitivity in T2DM mice.
Collapse
Affiliation(s)
- N Saleh
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| | - H E Elayan
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| | - M Zihlif
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| |
Collapse
|
160
|
Davis GR, Deville T, Guillory J, Bellar D, Nelson AG. Relationship between family history of type 2 diabetes and serum FGF21. Eur J Clin Invest 2017; 47:853-859. [PMID: 28881005 DOI: 10.1111/eci.12835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 09/02/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Determining predictive markers for the development of type 2 diabetes (T2D), particularly in young individuals, offers immense potential benefits in preventative medicine. Previous research examining serum fibroblast growth factor 21 (FGF21) in humans has revealed equivocal relationships with clinical markers of metabolic dysfunction. However, it is unknown to what extent, if any, first-degree family history of T2D (mother or father of the participant diagnosed with T2D) level affects serum FGF21 levels. The aim of this study was to determine whether in healthy individuals with FH+ (n = 18) and without FH- (n = 17) a family history of T2D affects serum FGF21. MATERIALS AND METHODS Fasting serum and clinical, metabolic and anthropometric measures were determined using a cross-sectional design. RESULTS Differences between groups for FGF21 were not significant (FH+ = 266 pg/mL ± 51·4, FH = 180 pg/mL ± 29; Z = 0·97, P = 0·33). Adiponectin values were lower in FH+ (8·81 μg/mL ± 2·14) compared to FH- (10·65 μg/mL ± 1·44; F = 8·83, P = 0·01). Resistin was negatively correlated with FGF21 for all participants (r = -0·38, P = 0·03), but no other clinical, metabolic, or serum markers were predictive for serum FGF21 in FH+ or FH-. CONCLUSIONS Serum FGF21 is not significantly different between FH+ and FH- in young, healthy individuals. Based upon the data of this pilot study, it is unclear whether serum FGF21 can be used as a stand-alone predictive marker for T2D in healthy subjects.
Collapse
Affiliation(s)
| | | | | | - David Bellar
- University of Louisiana at Lafayette, Lafayette, LA, USA
| | | |
Collapse
|
161
|
Newsom SA, Miller BF, Hamilton KL, Ehrlicher SE, Stierwalt HD, Robinson MM. Long-term rates of mitochondrial protein synthesis are increased in mouse skeletal muscle with high-fat feeding regardless of insulin-sensitizing treatment. Am J Physiol Endocrinol Metab 2017; 313:E552-E562. [PMID: 28698283 PMCID: PMC5792140 DOI: 10.1152/ajpendo.00144.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 11/22/2022]
Abstract
Skeletal muscle mitochondrial protein synthesis is regulated in part by insulin. The development of insulin resistance with diet-induced obesity may therefore contribute to impairments to protein synthesis and decreased mitochondrial respiration. Yet the impact of diet-induced obesity and insulin resistance on mitochondrial energetics is controversial, with reports varying from decreases to increases in mitochondrial respiration. We investigated the impact of changes in insulin sensitivity on long-term rates of mitochondrial protein synthesis as a mechanism for changes to mitochondrial respiration in skeletal muscle. Insulin resistance was induced in C57BL/6J mice using 4 wk of a high-fat compared with a low-fat diet. For 8 additional weeks, diets were enriched with pioglitazone to restore insulin sensitivity compared with nonenriched control low-fat or high-fat diets. Skeletal muscle mitochondrial protein synthesis was measured using deuterium oxide labeling during weeks 10-12 High-resolution respirometry was performed using palmitoyl-l-carnitine, glutamate+malate, and glutamate+malate+succinate as substrates for mitochondria isolated from quadriceps. Mitochondrial protein synthesis and palmitoyl- l-carnitine oxidation were increased in mice consuming a high-fat diet, regardless of differences in insulin sensitivity with pioglitazone treatment. There was no effect of diet or pioglitazone treatment on ADP-stimulated respiration or H2O2 emission using glutamate+malate or glutamate+malate+succinate. The results demonstrate no impairments to mitochondrial protein synthesis or respiration following induction of insulin resistance. Instead, mitochondrial protein synthesis was increased with a high-fat diet and may contribute to remodeling of the mitochondria to increase lipid oxidation capacity. Mitochondrial adaptations with a high-fat diet appear driven by nutrient availability, not intrinsic defects that contribute to insulin resistance.
Collapse
Affiliation(s)
- Sean A Newsom
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Benjamin F Miller
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, Colorado; and
| | - Karyn L Hamilton
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, Colorado; and
| | - Sarah E Ehrlicher
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Harrison D Stierwalt
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Matthew M Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon;
- Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
162
|
Frias FDT, Rocha KCE, de Mendonça M, Murata GM, Araujo HN, de Sousa LGO, de Sousa É, Hirabara SM, Leite NDC, Carneiro EM, Curi R, Silveira LR, Rodrigues AC. Fenofibrate reverses changes induced by high-fat diet on metabolism in mice muscle and visceral adipocytes. J Cell Physiol 2017; 233:3515-3528. [PMID: 28926107 DOI: 10.1002/jcp.26203] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023]
Abstract
The effect of fenofibrate on the metabolism of skeletal muscle and visceral white adipose tissue of diet-induced obese (DIO) mice was investigated. C57BL/6J male mice were fed either a control or high-fat diet for 8 weeks. Fenofibrate (50 mg/Kg BW, daily) was administered by oral gavage during the last two weeks of the experimental period. Insulin-stimulated glucose metabolism in soleus muscles, glucose tolerance test, insulin tolerance test, indirect calorimetry, lipolysis of visceral white adipose tissue, expression of miR-103-3p in adipose tissue, and miR-1a, miR-133a/b, miR-206, let7b-5p, miR-23b-3p, miR-29-3p, miR-143-3p in soleus muscle, genes related to glucose and fatty acid metabolism in adipose tissue and soleus muscle, and proteins (phospho-AMPKα2, Pgc1α, Cpt1b), intramuscular lipid staining, and activities of fatty acid oxidation enzymes in skeletal muscle were investigated. In DIO mice, fenofibrate prevented weight gain induced by HFD feeding by increasing energy expenditure; improved whole body glucose homeostasis, and in skeletal muscle, increased insulin dependent glucose uptake, miR-1a levels, reduced intramuscular lipid accumulation, and phospho-AMPKα2 levels. In visceral adipose tissue of obese mice, fenofibrate decreased basal lipolysis rate and visceral adipocytes hypertrophy, and induced the expression of Glut-4, Irs1, and Cav-1 mRNA and miR-103-3p suggesting a higher insulin sensitivity of the adipocytes. The evidence is presented herein that beneficial effects of fenofibrate on body weight, glucose homeostasis, and muscle metabolism might be related to its action in adipose tissue. Moreover, fenofibrate regulates miR-1a-3p in soleus and miR-103-3p in adipose tissue, suggesting these microRNAs might contribute to fenofibrate beneficial effects on metabolism.
Collapse
Affiliation(s)
- Flávia de T Frias
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Karina C E Rocha
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Mariana de Mendonça
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Gilson M Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Luís G O de Sousa
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Érica de Sousa
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Sandro M Hirabara
- Interdisciplinary Post-Graduate Program in Health Science, Cruzeiro do Sul University, Sao Paulo, Brazil
| | - Nayara de C Leite
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,Interdisciplinary Post-Graduate Program in Health Science, Cruzeiro do Sul University, Sao Paulo, Brazil
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, Campinas, Sao Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Alice C Rodrigues
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
163
|
Kim K, Ahn N, Jung S, Park S. Effects of intermittent ladder-climbing exercise training on mitochondrial biogenesis and endoplasmic reticulum stress of the cardiac muscle in obese middle-aged rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:633-641. [PMID: 29200906 PMCID: PMC5709480 DOI: 10.4196/kjpp.2017.21.6.633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
The aim of this study is to investigate the effects of intermittent ladder-climbing exercise training on mitochondrial biogenesis and ER stress of the cardiac muscle in high fat diet-induced obese middle-aged rats. We induced obesity over 6 weeks of period in 40 male Sprague-Dawley rats around 50 weeks old, and were randomly divided into four experimental groups: chow, HFD, exercise+HFD, and exercise+chow. The exercising groups underwent high-intensity intermittent training using a ladder-climbing and weight exercise 3 days/week for a total of 8 weeks. High-fat diet and concurrent exercise resulted in no significant reduction in body weight but caused a significant reduction in visceral fat weight (p<0.05). Expression of PPARδ increased in the exercise groups and was significantly increased in the high-fat diet+exercise group (p<0.05). Among the ER stress-related proteins, the expression levels of p-PERK and CHOP, related to cardiac muscle damage, were significantly higher in the cardiac muscle of the high-fat diet group (p<0.05), and were significantly reduced by intermittent ladder-climbing exercise training (p<0.05). Specifically, this reduction was greater when the rats underwent exercise after switching back to the chow diet with a reduced caloric intake. Collectively, these results suggest that the combination of intermittent ladder-climbing exercise training and a reduced caloric intake can decrease the levels of ER stress-related proteins that contribute to cardiac muscle damage in obesity and aging. However, additional validation is required to understand the effects of these changes on mitochondrial biogenesis during exercise.
Collapse
Affiliation(s)
- Kijin Kim
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea
| | - Nayoung Ahn
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea
| | - Suryun Jung
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea
| | - Solee Park
- Department of Physical Education, College of Physical Education, Keimyung University, Daegu 42601, Korea
| |
Collapse
|
164
|
Pataky MW, Wang H, Yu CS, Arias EB, Ploutz-Snyder RJ, Zheng X, Cartee GD. High-Fat Diet-Induced Insulin Resistance in Single Skeletal Muscle Fibers is Fiber Type Selective. Sci Rep 2017; 7:13642. [PMID: 29057943 PMCID: PMC5651812 DOI: 10.1038/s41598-017-12682-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/18/2017] [Indexed: 11/17/2022] Open
Abstract
Skeletal muscle is the major site for insulin-stimulated glucose disposal, and muscle insulin resistance confers many negative health outcomes. Muscle is composed of multiple fiber types, and conventional analysis of whole muscles cannot elucidate fiber type differences at the cellular level. Previous research demonstrated that a brief (two weeks) high fat diet (HFD) caused insulin resistance in rat skeletal muscle. The primary aim of this study was to determine in rat skeletal muscle the influence of a brief (two weeks) HFD on glucose uptake (GU) ± insulin in single fibers that were also characterized for fiber type. Epitrochlearis muscles were incubated with [3H]-2-deoxyglucose (2DG) ± 100 µU/ml insulin. Fiber type (myosin heavy chain expression) and 2DG accumulation were measured in whole muscles and single fibers. Although fiber type composition of whole muscles did not differ between diet groups, GU of insulin-stimulated whole muscles from LFD rats significantly exceeded HFD values (P < 0.005). For HFD versus LFD rats, GU of insulin-stimulated single fibers was significantly (P < 0.05) lower for IIA, IIAX, IIBX, IIB, and approached significance for IIX (P = 0.100), but not type I (P = 0.776) fibers. These results revealed HFD-induced insulin resistance was attributable to fiber type selective insulin resistance and independent of altered fiber type composition.
Collapse
Affiliation(s)
- Mark W Pataky
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Carmen S Yu
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Xiaohua Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA. .,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA. .,Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
165
|
Pinho RA, Sepa-Kishi DM, Bikopoulos G, Wu MV, Uthayakumar A, Mohasses A, Hughes MC, Perry CGR, Ceddia RB. High-fat diet induces skeletal muscle oxidative stress in a fiber type-dependent manner in rats. Free Radic Biol Med 2017; 110:381-389. [PMID: 28690197 DOI: 10.1016/j.freeradbiomed.2017.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/15/2017] [Accepted: 07/05/2017] [Indexed: 02/02/2023]
Abstract
This study investigated the effects of high-fat (HF) diet on parameters of oxidative stress among muscles with distinct fiber type composition and oxidative capacities. To accomplish that, male Wistar rats were fed either a low-fat standard chow (SC) or a HF diet for 8 weeks. Soleus, extensor digitorum longus (EDL), and epitrochlearis muscles were collected and mitochondrial H2O2 (mtH2O2) emission, palmitate oxidation, and gene expression and antioxidant system were measured. Chronic HF feeding enhanced fat oxidation in oxidative and glycolytic muscles. It also caused a significant reduction in mtH2O2 emission in the EDL muscle, although a tendency towards a reduction was also found in the soleus and epitrochlearis muscles. In the epitrochlearis, HF diet increased mRNA expression of the NADPH oxidase complex; however, this muscle also showed an increase in the expression of antioxidant proteins, suggesting a higher capacity to generate and buffer ROS. The soleus muscle, despite being highly oxidative, elicited H2O2 emission rates equivalent to only 20% and 35% of the values obtained for EDL and epitrochlearis muscles, respectively. Furthermore, the Epi muscle with the lowest oxidative capacity was the second highest in H2O2 emission. In conclusion, it appears that intrinsic differences related to the distribution of type I and type II fibers, rather than oxidative capacity, drove the activity of the anti- and pro-oxidant systems and determine ROS production in different skeletal muscles. This also suggests that the impact of potentially deleterious effects of ROS production on skeletal muscle metabolism/function under lipotoxic conditions is fiber type-specific.
Collapse
Affiliation(s)
- Ricardo A Pinho
- Laboratory of Exercise Biochemistry and Physiology, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma (UNESC), Santa Catarina, Brazil.
| | - Diane M Sepa-Kishi
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - George Bikopoulos
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Michelle V Wu
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Abinas Uthayakumar
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Arta Mohasses
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Meghan C Hughes
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Rolando B Ceddia
- School of Kinesiology and Health Science - Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
166
|
Vartanian V, Tumova J, Dobrzyn P, Dobrzyn A, Nakabeppu Y, Lloyd RS, Sampath H. 8-oxoguanine DNA glycosylase (OGG1) deficiency elicits coordinated changes in lipid and mitochondrial metabolism in muscle. PLoS One 2017; 12:e0181687. [PMID: 28727777 PMCID: PMC5519207 DOI: 10.1371/journal.pone.0181687] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 07/04/2017] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress resulting from endogenous and exogenous sources causes damage to cellular components, including genomic and mitochondrial DNA. Oxidative DNA damage is primarily repaired via the base excision repair pathway that is initiated by DNA glycosylases. 8-oxoguanine DNA glycosylase (OGG1) recognizes and cleaves oxidized and ring-fragmented purines, including 8-oxoguanine, the most commonly formed oxidative DNA lesion. Mice lacking the OGG1 gene product are prone to multiple features of the metabolic syndrome, including high-fat diet-induced obesity, hepatic steatosis, and insulin resistance. Here, we report that OGG1-deficient mice also display skeletal muscle pathologies, including increased muscle lipid deposition and alterations in genes regulating lipid uptake and mitochondrial fission in skeletal muscle. In addition, expression of genes of the TCA cycle and of carbohydrate and lipid metabolism are also significantly altered in muscle of OGG1-deficient mice. These tissue changes are accompanied by marked reductions in markers of muscle function in OGG1-deficient animals, including decreased grip strength and treadmill endurance. Collectively, these data indicate a role for skeletal muscle OGG1 in the maintenance of optimal tissue function.
Collapse
Affiliation(s)
- Vladimir Vartanian
- From the Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jana Tumova
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Pawel Dobrzyn
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - R. Stephen Lloyd
- From the Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Harini Sampath
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, United States of America
- Rutgers Center for Lipid Research and Center for Digestive Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, United States of America
| |
Collapse
|
167
|
Franko A, Kunze A, Böse M, von Kleist-Retzow JC, Paulsson M, Hartmann U, Wiesner RJ. Impaired Insulin Signaling is Associated with Hepatic Mitochondrial Dysfunction in IR +/--IRS-1 +/- Double Heterozygous (IR-IRS1dh) Mice. Int J Mol Sci 2017; 18:ijms18061156. [PMID: 28556799 PMCID: PMC5485980 DOI: 10.3390/ijms18061156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/21/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Mitochondria play a pivotal role in energy metabolism, but whether insulin signaling per se could regulate mitochondrial function has not been identified yet. To investigate whether mitochondrial function is regulated by insulin signaling, we analyzed muscle and liver of insulin receptor (IR)+/−-insulin receptor substrate-1 (IRS-1)+/− double heterozygous (IR-IRS1dh) mice, a well described model for insulin resistance. IR-IRS1dh mice were studied at the age of 6 and 12 months and glucose metabolism was determined by glucose and insulin tolerance tests. Mitochondrial enzyme activities, oxygen consumption, and membrane potential were assessed using spectrophotometric, respirometric, and proton motive force analysis, respectively. IR-IRS1dh mice showed elevated serum insulin levels. Hepatic mitochondrial oxygen consumption was reduced in IR-IRS1dh animals at 12 months of age. Furthermore, 6-month-old IR-IRS1dh mice demonstrated enhanced mitochondrial respiration in skeletal muscle, but a tendency of impaired glucose tolerance. On the other hand, 12-month-old IR-IRS1dh mice showed improved glucose tolerance, but normal muscle mitochondrial function. Our data revealed that deficiency in IR/IRS-1 resulted in normal or even elevated skeletal muscle, but impaired hepatic mitochondrial function, suggesting a direct cross-talk between insulin signaling and mitochondria in the liver.
Collapse
Affiliation(s)
- Andras Franko
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str. 10, D-72076 Tübingen, Germany.
| | - Alexander Kunze
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
| | - Marlen Böse
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
| | - Jürgen-Christoph von Kleist-Retzow
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Department of Pediatrics, University Hospital Cologne, Kerpener Str. 62, D-50937 Cologne, Germany.
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Köln, D-50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Köln, D-50931 Cologne, Germany.
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Köln, Joseph-Stelzmann-Str. 52, D-50931 Cologne, Germany.
| | - Rudolf J Wiesner
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Robert-Koch-Str. 39, D-50931 Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Köln, D-50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Köln, D-50931 Cologne, Germany.
| |
Collapse
|
168
|
Callahan ZJ, Oxendine MJ, Schaeffer PJ. Intramuscular triglyceride content precedes impaired glucose metabolism without evidence for mitochondrial dysfunction during early development of a diabetic phenotype. Appl Physiol Nutr Metab 2017; 42:963-972. [PMID: 28538106 DOI: 10.1139/apnm-2016-0685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The incidence of type 2 diabetes is highly correlated with obesity; however, there is a lack of research elucidating the temporal progression. Transgenic FVB/N UCP-dta mice, which develop a diabetic phenotype, and their nontransgenic littermates were fed either a high-fat or normal-chow diet and were studied at 6, 9, 12, 15, 18, 21, and 24 weeks of age to test the hypothesis that increased lipid accumulation in skeletal muscle causes mitochondrial dysfunction, leading to the development of insulin resistance. Body composition, intramuscular triglyceride (IMTG) content, glucose metabolism, and mitochondrial function were measured to determine if IMTG drove mitochondrial dysfunction, leading to the development of type 2 diabetes. High-fat-fed transgenic mice had a significantly greater body mass, lipid mass, and IMTG content beginning early in the experiment. Glucose tolerance tests revealed that high-fat-fed transgenic mice developed a significantly insulin resistant response compared with the other 3 groups toward the end of the time course while plasma insulin was elevated very early in the time course. There was no significant difference in several measures of metabolic function throughout the time course. Long-term high-fat feeding in transgenic mice produced increases in IMTG, adiposity, body mass, and plasma insulin accompanied by decreases in glucose metabolism, but did not reveal any deficits in mitochondrial function or regulation during the early stage of the development of type 2 diabetes. It does not appear that lipotoxicity is driving defects in mitochondrial function prior to the onset of insulin resistance.
Collapse
Affiliation(s)
- Zachary J Callahan
- Department of Biology, Miami University, Oxford, OH 45056, USA.,Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Michael J Oxendine
- Department of Biology, Miami University, Oxford, OH 45056, USA.,Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Paul J Schaeffer
- Department of Biology, Miami University, Oxford, OH 45056, USA.,Department of Biology, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
169
|
Boulinguiez A, Staels B, Duez H, Lancel S. Mitochondria and endoplasmic reticulum: Targets for a better insulin sensitivity in skeletal muscle? Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:901-916. [PMID: 28529179 DOI: 10.1016/j.bbalip.2017.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/16/2022]
Abstract
Obesity and its associated metabolic disorders represent a major health burden, with economic and social consequences. Although adapted lifestyle and bariatric surgery are effective in reducing body weight, obesity prevalence is still rising. Obese individuals often become insulin-resistant. Obesity impacts on insulin responsive organs, such as the liver, adipose tissue and skeletal muscle, and increases the risk of cardiovascular diseases, type 2 diabetes and cancer. In this review, we discuss the effects of obesity and insulin resistance on skeletal muscle, an important organ for the control of postprandial glucose. The roles of mitochondria and the endoplasmic reticulum in insulin signaling are highlighted and potential innovative research and treatment perspectives are proposed.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| |
Collapse
|
170
|
Koh JH, Hancock CR, Terada S, Higashida K, Holloszy JO, Han DH. PPARβ Is Essential for Maintaining Normal Levels of PGC-1α and Mitochondria and for the Increase in Muscle Mitochondria Induced by Exercise. Cell Metab 2017; 25:1176-1185.e5. [PMID: 28467933 PMCID: PMC5894349 DOI: 10.1016/j.cmet.2017.04.029] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 01/24/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022]
Abstract
The objective of this study was to evaluate the specific mechanism(s) by which PPARβ regulates mitochondrial content in skeletal muscle. We discovered that PPARβ increases PGC-1α by protecting it from degradation by binding to PGC-1α and limiting ubiquitination. PPARβ also induces an increase in nuclear respiratory factor 1 (NRF-1) expression, resulting in increases in mitochondrial respiratory chain proteins and MEF2A, for which NRF-1 is a transcription factor. There was also an increase in AMP kinase phosphorylation mediated by an NRF-1-induced increase in CAM kinase kinase-β (CaMKKβ). Knockdown of PPARβ resulted in large decreases in the levels of PGC-1α and mitochondrial proteins and a marked attenuation of the exercise-induced increase in mitochondrial biogenesis. In conclusion, PPARβ induces an increase in PGC-1α protein, and PPARβ is a transcription factor for NRF-1. Thus, PPARβ plays essential roles in the maintenance and adaptive increase in mitochondrial enzymes in skeletal muscle by exercise.
Collapse
Affiliation(s)
- Jin-Ho Koh
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chad R Hancock
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shin Terada
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kazuhiko Higashida
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John O Holloszy
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Dong-Ho Han
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
171
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
172
|
López-Lluch G. Mitochondrial activity and dynamics changes regarding metabolism in ageing and obesity. Mech Ageing Dev 2017; 162:108-121. [DOI: 10.1016/j.mad.2016.12.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/24/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
|
173
|
Mannerås-Holm L, Schönke M, Brozinick JT, Vetterli L, Bui HH, Sanders P, Nascimento EBM, Björnholm M, Chibalin AV, Zierath JR. Diacylglycerol kinase ε deficiency preserves glucose tolerance and modulates lipid metabolism in obese mice. J Lipid Res 2017; 58:907-915. [PMID: 28246337 DOI: 10.1194/jlr.m074443] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/13/2017] [Indexed: 12/19/2022] Open
Abstract
Diacylglycerol kinases (DGKs) catalyze the phosphorylation and conversion of diacylglycerol (DAG) into phosphatidic acid. DGK isozymes have unique primary structures, expression patterns, subcellular localizations, regulatory mechanisms, and DAG preferences. DGKε has a hydrophobic segment that promotes its attachment to membranes and shows substrate specificity for DAG with an arachidonoyl acyl chain in the sn-2 position of the substrate. We determined the role of DGKε in the regulation of energy and glucose homeostasis in relation to diet-induced insulin resistance and obesity using DGKε-KO and wild-type mice. Lipidomic analysis revealed elevated unsaturated and saturated DAG species in skeletal muscle of DGKε KO mice, which was paradoxically associated with increased glucose tolerance. Although skeletal muscle insulin sensitivity was unaltered, whole-body respiratory exchange ratio was reduced, and abundance of mitochondrial markers was increased, indicating a greater reliance on fat oxidation and intracellular lipid metabolism in DGKε KO mice. Thus, the increased intracellular lipids in skeletal muscle from DGKε KO mice may undergo rapid turnover because of increased mitochondrial function and lipid oxidation, rather than storage, which in turn may preserve insulin sensitivity. In conclusion, DGKε plays a role in glucose and energy homeostasis by modulating lipid metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Louise Mannerås-Holm
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Milena Schönke
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Laurène Vetterli
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hai-Hoang Bui
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Philip Sanders
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Emmani B M Nascimento
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Björnholm
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
174
|
Barbosa de Queiroz K, Honorato-Sampaio K, Rossoni Júnior JV, Andrade Leal D, Pinto ABG, Kappes-Becker L, Evangelista EA, Guerra-Sá R. Physical activity prevents alterations in mitochondrial ultrastructure and glucometabolic parameters in a high-sugar diet model. PLoS One 2017; 12:e0172103. [PMID: 28199417 PMCID: PMC5310863 DOI: 10.1371/journal.pone.0172103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/31/2017] [Indexed: 11/19/2022] Open
Abstract
Endurance exercise is a remarkable intervention for the treatment of many diseases. Mitochondrial changes on skeletal muscle are likely important for many of the benefits provided by exercise. In this study, we aimed to evaluate the effects that a regular physical activity (swimming without workload) has on mitochondrial morphological alterations and glucometabolic parameters induced by a high-sugar diet (HSD). Weaned male Wistar rats fed with a standard diet or a HSD (68% carbohydrate) were subjected to 60 minutes of regular physical activity by swimming (without workload) for four- (20 sessions) or eight-week (40 sessions) periods. After training, animals were euthanized and the sera, adipose tissues, and skeletal muscles were collected for further analysis. The HSD increased body weight after an 8-week period; it also increased the fat pads and the adipose index, resulting in glucose intolerance and insulin resistance (IR). Transmission electron microscopy showed an increase in alterations of mitochondrial ultrastructure in the gastrocnemius muscle, as well as a decrease in superoxide dismutase (SOD) activity, and an increase in protein carbonylation. Regular physical activity partially reverted these alterations in rats fed a HSD, preventing mitochondrial morphological alterations and IR. Moreover, we observed a decrease in Pgc1α expression (qPCR analysis) in STD-EXE group and a less pronounced reduction in HSD-EXE group after an 8-week period. Thus, regular physical activity (swimming without workload) in rats fed a HSD can prevent mitochondrial dysfunction and IR, highlighting the crucial role for physical activity on metabolic homeostasis.
Collapse
Affiliation(s)
- Karina Barbosa de Queiroz
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
- * E-mail:
| | - Kinulpe Honorato-Sampaio
- Faculdade de Medicina, Campus JK, Universidade Federal dos Vales Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brasil
| | - Joamyr Victor Rossoni Júnior
- Laboratório de Bioquímica Metabólica, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Diego Andrade Leal
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | | | - Lenice Kappes-Becker
- Centro de Esportes, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Elisio Alberto Evangelista
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Renata Guerra-Sá
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| |
Collapse
|
175
|
Cholesterol contributes to dopamine-neuronal loss in MPTP mouse model of Parkinson's disease: Involvement of mitochondrial dysfunctions and oxidative stress. PLoS One 2017; 12:e0171285. [PMID: 28170429 PMCID: PMC5295696 DOI: 10.1371/journal.pone.0171285] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/19/2017] [Indexed: 01/24/2023] Open
Abstract
Hypercholesterolemia is a known contributor to the pathogenesis of Alzheimer’s disease while its role in the occurrence of Parkinson’s disease (PD) is only conjecture and far from conclusive. Altered antioxidant homeostasis and mitochondrial functions are the key mechanisms in loss of dopaminergic neurons in the substantia nigra (SN) region of the midbrain in PD. Hypercholesterolemia is reported to cause oxidative stress and mitochondrial dysfunctions in the cortex and hippocampus regions of the brain in rodents. However, the impact of hypercholesterolemia on the midbrain dopaminergic neurons in animal models of PD remains elusive. We tested the hypothesis that hypercholesterolemia in MPTP model of PD would potentiate dopaminergic neuron loss in SN by disrupting mitochondrial functions and antioxidant homeostasis. It is evident from the present study that hypercholesterolemia in naïve animals caused dopamine neuronal loss in SN with subsequent reduction in striatal dopamine levels producing motor impairment. Moreover, in the MPTP model of PD, hypercholesterolemia exacerbated MPTP-induced reduction of striatal dopamine as well as dopaminergic neurons in SN with motor behavioral depreciation. Activity of mitochondrial complexes, mainly complex-I and III, was impaired severely in the nigrostriatal pathway of hypercholesterolemic animals treated with MPTP. Hypercholesterolemia caused oxidative stress in the nigrostriatal pathway with increased generation of hydroxyl radicals and enhanced activity of antioxidant enzymes, which were further aggravated in the hypercholesterolemic mice with Parkinsonism. In conclusion, our findings provide evidence of increased vulnerability of the midbrain dopaminergic neurons in PD with hypercholesterolemia.
Collapse
|
176
|
Gonzalez-Franquesa A, Patti ME. Insulin Resistance and Mitochondrial Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:465-520. [DOI: 10.1007/978-3-319-55330-6_25] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
177
|
Tamura Y, Hatta H. Heat stress induces mitochondrial adaptations in skeletal muscle. ACTA ACUST UNITED AC 2017. [DOI: 10.7600/jpfsm.6.151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yuki Tamura
- Department of Exercise Physiology, Nippon Sport Science University
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University
| | - Hideo Hatta
- Department of Sports Sciences, The University of Tokyo
| |
Collapse
|
178
|
Reynés B, Palou M, Palou A. Gene expression modulation of lipid and central energetic metabolism related genes by high-fat diet intake in the main homeostatic tissues. Food Funct 2017; 8:629-650. [DOI: 10.1039/c6fo01473a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
HF diet feeding affects the energy balance by transcriptional metabolic adaptations, based in direct gene expression modulation, perinatal programing and transcriptional factor regulation, which could be affected by the animal model, gender or period of dietary treatment.
Collapse
Affiliation(s)
- Bàrbara Reynés
- Laboratory of Molecular Biology
- Nutrition and Biotechnology
- Universitat de les Illes Balears and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn)
- Palma de Mallorca
- Spain
| | - Mariona Palou
- Alimentómica SL (Spin off no. 001 from UIB)
- Palma Mallorca
- Spain
| | - Andreu Palou
- Laboratory of Molecular Biology
- Nutrition and Biotechnology
- Universitat de les Illes Balears and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn)
- Palma de Mallorca
- Spain
| |
Collapse
|
179
|
Laurens C, Bourlier V, Mairal A, Louche K, Badin PM, Mouisel E, Montagner A, Marette A, Tremblay A, Weisnagel JS, Guillou H, Langin D, Joanisse DR, Moro C. Perilipin 5 fine-tunes lipid oxidation to metabolic demand and protects against lipotoxicity in skeletal muscle. Sci Rep 2016; 6:38310. [PMID: 27922115 PMCID: PMC5138838 DOI: 10.1038/srep38310] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/07/2016] [Indexed: 11/09/2022] Open
Abstract
Lipid droplets (LD) play a central role in lipid homeostasis by controlling transient fatty acid (FA) storage and release from triacylglycerols stores, while preventing high levels of cellular toxic lipids. This crucial function in oxidative tissues is altered in obesity and type 2 diabetes. Perilipin 5 (PLIN5) is a LD protein whose mechanistic and causal link with lipotoxicity and insulin resistance has raised controversies. We investigated here the physiological role of PLIN5 in skeletal muscle upon various metabolic challenges. We show that PLIN5 protein is elevated in endurance-trained (ET) subjects and correlates with muscle oxidative capacity and whole-body insulin sensitivity. When overexpressed in human skeletal muscle cells to recapitulate the ET phenotype, PLIN5 diminishes lipolysis and FA oxidation under basal condition, but paradoxically enhances FA oxidation during forskolin- and contraction- mediated lipolysis. Moreover, PLIN5 partly protects muscle cells against lipid-induced lipotoxicity. In addition, we demonstrate that down-regulation of PLIN5 in skeletal muscle inhibits insulin-mediated glucose uptake under normal chow feeding condition, while paradoxically improving insulin sensitivity upon high-fat feeding. These data highlight a key role of PLIN5 in LD function, first by finely adjusting LD FA supply to mitochondrial oxidation, and second acting as a protective factor against lipotoxicity in skeletal muscle.
Collapse
Affiliation(s)
- Claire Laurens
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Virginie Bourlier
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Aline Mairal
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Katie Louche
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Pierre-Marie Badin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Etienne Mouisel
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| | - Alexandra Montagner
- University of Toulouse, Paul Sabatier University, France.,INRA, UMR 1331, TOXALIM, Toulouse, France
| | - André Marette
- Department of Medicine, Laval University, Quebec City, Canada.,Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada
| | - Angelo Tremblay
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada.,Department of Kinesiology, Laval University, Quebec City, Canada
| | | | - Hervé Guillou
- University of Toulouse, Paul Sabatier University, France.,INRA, UMR 1331, TOXALIM, Toulouse, France
| | - Dominique Langin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France.,Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Denis R Joanisse
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada.,Department of Kinesiology, Laval University, Quebec City, Canada
| | - Cedric Moro
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, France
| |
Collapse
|
180
|
A-type procyanidins from litchi pericarp ameliorate hyperglycaemia by regulating hepatic and muscle glucose metabolism in streptozotocin (STZ)-induced diabetic mice fed with high fat diet. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.08.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
181
|
Hyatt HW, Kephart WC, Holland AM, Mumford P, Mobley CB, Lowery RP, Roberts MD, Wilson JM, Kavazis AN. A Ketogenic Diet in Rodents Elicits Improved Mitochondrial Adaptations in Response to Resistance Exercise Training Compared to an Isocaloric Western Diet. Front Physiol 2016; 7:533. [PMID: 27877138 PMCID: PMC5099251 DOI: 10.3389/fphys.2016.00533] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/26/2016] [Indexed: 01/16/2023] Open
Abstract
Purpose: Ketogenic diets (KD) can facilitate weight loss, but their effects on skeletal muscle remain equivocal. In this experiment we investigated the effects of two diets on skeletal muscle mitochondrial coupling, mitochondrial complex activity, markers of oxidative stress, and gene expression in sedentary and resistance exercised rats. Methods: Male Sprague-Dawley rats (9-10 weeks of age, 300-325 g) were fed isocaloric amounts of either a KD (17 g/day, 5.2 kcal/g, 20.2% protein, 10.3% CHO, 69.5% fat, n = 16) or a Western diet (WD) (20 g/day, 4.5 kcal/g, 15.2% protein, 42.7% CHO, 42.0% fat, n = 16) for 6 weeks. During these 6 weeks animals were either sedentary (SED, n = 8 per diet group) or voluntarily exercised using resistance-loaded running wheels (EXE, n = 8 per diet group). Gastrocnemius was excised and used for mitochondrial isolation and biochemical analyses. Results: In the presence of a complex II substrate, the respiratory control ratio (RCR) of isolated gastrocnemius mitochondria was higher (p < 0.05) in animals fed the KD compared to animals fed the WD. Complex I and IV enzyme activity was higher (p < 0.05) in EXE animals regardless of diet. SOD2 protein levels and GLUT4 and PGC1α mRNA expression were higher (p < 0.05) in EXE animals regardless of diet. Conclusion: Our data indicate that skeletal muscle mitochondrial coupling of complex II substrates is more efficient in chronically resistance trained rodents fed a KD. These findings may provide merit for further investigation, perhaps on humans.
Collapse
Affiliation(s)
- Hayden W Hyatt
- School of Kinesiology, Auburn University Auburn, AL, USA
| | | | | | - Petey Mumford
- School of Kinesiology, Auburn University Auburn, AL, USA
| | | | - Ryan P Lowery
- Department of Health Sciences and Human Performance, University of Tampa Tampa, FL, USA
| | | | - Jacob M Wilson
- Department of Health Sciences and Human Performance, University of Tampa Tampa, FL, USA
| | | |
Collapse
|
182
|
Hesselink MKC, Schrauwen-Hinderling V, Schrauwen P. Skeletal muscle mitochondria as a target to prevent or treat type 2 diabetes mellitus. Nat Rev Endocrinol 2016; 12:633-645. [PMID: 27448057 DOI: 10.1038/nrendo.2016.104] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Low levels of physical activity and the presence of obesity are associated with mitochondrial dysfunction. In addition, mitochondrial dysfunction has been associated with the development of insulin resistance and type 2 diabetes mellitus (T2DM). Although the evidence for a causal relationship between mitochondrial function and insulin resistance is still weak, emerging evidence indicates that boosting mitochondrial function might be beneficial to patient health. Exercise training is probably the most recognized promoter of mitochondrial function and insulin sensitivity and hence is still regarded as the best strategy to prevent and treat T2DM. Animal data, however, have revealed several new insights into the regulation of mitochondrial metabolism, and novel targets for interventions to boost mitochondrial function have emerged. Importantly, many of these targets seem to be regulated by factors such as nutrition, ambient temperature and circadian rhythms, which provides a basis for nonpharmacological strategies to prevent or treat T2DM in humans. Here, we will review the current evidence that mitochondrial function can be targeted therapeutically to improve insulin sensitivity and to prevent T2DM, focusing mainly on human intervention studies.
Collapse
Affiliation(s)
- Matthijs K C Hesselink
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- Department of Radiology, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX, Maastricht, Netherlands
| | - Patrick Schrauwen
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| |
Collapse
|
183
|
Brace LE, Vose SC, Stanya K, Gathungu RM, Marur VR, Longchamp A, Treviño-Villarreal H, Mejia P, Vargas D, Inouye K, Bronson RT, Lee CH, Neilan E, Kristal BS, Mitchell JR. Increased oxidative phosphorylation in response to acute and chronic DNA damage. NPJ Aging Mech Dis 2016; 2:16022. [PMID: 28721274 PMCID: PMC5514997 DOI: 10.1038/npjamd.2016.22] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 06/11/2016] [Accepted: 07/25/2016] [Indexed: 12/25/2022] Open
Abstract
Accumulation of DNA damage is intricately linked to aging, aging-related diseases and progeroid syndromes such as Cockayne syndrome (CS). Free radicals from endogenous oxidative energy metabolism can damage DNA, however the potential of acute or chronic DNA damage to modulate cellular and/or organismal energy metabolism remains largely unexplored. We modeled chronic endogenous genotoxic stress using a DNA repair-deficient Csa-/-|Xpa-/- mouse model of CS. Exogenous genotoxic stress was modeled in mice in vivo and primary cells in vitro treated with different genotoxins giving rise to diverse spectrums of lesions, including ultraviolet radiation, intrastrand crosslinking agents and ionizing radiation. Both chronic endogenous and acute exogenous genotoxic stress increased mitochondrial fatty acid oxidation (FAO) on the organismal level, manifested by increased oxygen consumption, reduced respiratory exchange ratio, progressive adipose loss and increased FAO in tissues ex vivo. In multiple primary cell types, the metabolic response to different genotoxins manifested as a cell-autonomous increase in oxidative phosphorylation (OXPHOS) subsequent to a transient decline in steady-state NAD+ and ATP levels, and required the DNA damage sensor PARP-1 and energy-sensing kinase AMPK. We conclude that increased FAO/OXPHOS is a general, beneficial, adaptive response to DNA damage on cellular and organismal levels, illustrating a fundamental link between genotoxic stress and energy metabolism driven by the energetic cost of DNA damage. Our study points to therapeutic opportunities to mitigate detrimental effects of DNA damage on primary cells in the context of radio/chemotherapy or progeroid syndromes.
Collapse
Affiliation(s)
- Lear E Brace
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sarah C Vose
- Division of Environmental Health, Vermont Department of Health, Burlington, VT, USA
| | - Kristopher Stanya
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Rose M Gathungu
- Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Vasant R Marur
- Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | - Pedro Mejia
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Dorathy Vargas
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Karen Inouye
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Roderick T Bronson
- Rodent Histopathology Core, Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward Neilan
- Genetics and Metabolism Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce S Kristal
- Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
184
|
Characterization of the role of sphingomyelin synthase 2 in glucose metabolism in whole-body and peripheral tissues in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:688-702. [DOI: 10.1016/j.bbalip.2016.04.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/20/2016] [Accepted: 04/30/2016] [Indexed: 11/22/2022]
|
185
|
Heden TD, Neufer PD, Funai K. Looking Beyond Structure: Membrane Phospholipids of Skeletal Muscle Mitochondria. Trends Endocrinol Metab 2016; 27:553-562. [PMID: 27370525 PMCID: PMC4958499 DOI: 10.1016/j.tem.2016.05.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/23/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022]
Abstract
Skeletal muscle mitochondria are highly dynamic and are capable of tremendous expansion to meet cellular energetic demands. Such proliferation in mitochondrial mass requires a synchronized supply of enzymes and structural phospholipids. While transcriptional regulation of mitochondrial enzymes has been extensively studied, there is limited information on how mitochondrial membrane lipids are generated in skeletal muscle. Herein we describe how each class of phospholipids that constitute mitochondrial membranes are synthesized and/or imported, and summarize genetic evidence indicating that membrane phospholipid composition represents a significant modulator of skeletal muscle mitochondrial respiratory function. We also discuss how skeletal muscle mitochondrial phospholipids may mediate the effect of diet and exercise on oxidative metabolism.
Collapse
Affiliation(s)
- Timothy D Heden
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Katsuhiko Funai
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA; Department of Kinesiology, East Carolina University, Greenville, NC, USA; Department of Physiology, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
186
|
Affourtit C. Mitochondrial involvement in skeletal muscle insulin resistance: A case of imbalanced bioenergetics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1678-93. [PMID: 27473535 DOI: 10.1016/j.bbabio.2016.07.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/19/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
Skeletal muscle insulin resistance in obesity associates with mitochondrial dysfunction, but the causality of this association is controversial. This review evaluates mitochondrial models of nutrient-induced muscle insulin resistance. It transpires that all models predict that insulin resistance arises as a result of imbalanced cellular bioenergetics. The nature and precise origin of the proposed insulin-numbing molecules differ between models but all species only accumulate when metabolic fuel supply outweighs energy demand. This observation suggests that mitochondrial deficiency in muscle insulin resistance is not merely owing to intrinsic functional defects, but could instead be an adaptation to nutrient-induced changes in energy expenditure. Such adaptive effects are likely because muscle ATP supply is fully driven by energy demand. This market-economic control of myocellular bioenergetics offers a mechanism by which insulin-signalling deficiency can cause apparent mitochondrial dysfunction, as insulin resistance lowers skeletal muscle anabolism and thus dampens ATP demand and, consequently, oxidative ATP synthesis.
Collapse
Affiliation(s)
- Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth University, Drake Circus, PL4 8AA Plymouth, UK.
| |
Collapse
|
187
|
Mizunoya W, Okamoto S, Miyahara H, Akahoshi M, Suzuki T, Do MKQ, Ohtsubo H, Komiya Y, Qahar M, Waga T, Nakazato K, Ikeuchi Y, Anderson JE, Tatsumi R. Fast-to-slow shift of muscle fiber-type composition by dietary apple polyphenols in rats: Impact of the low-dose supplementation. Anim Sci J 2016; 88:489-499. [PMID: 27417667 DOI: 10.1111/asj.12655] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 03/27/2016] [Accepted: 04/07/2016] [Indexed: 11/30/2022]
Abstract
Our previous studies demonstrated that an 8-week intake of 5% (w/w) apple polyphenol (APP) in the diet improves muscle endurance of young-adult rats. In order to identify a lower limit of the dietary contribution of APP to the effect, the experiments were designed for lower-dose supplementation (8-week feeding of 0.5% APP in AIN-93G diet) to 12-week-old male Sprague-Dawley rats. Results clearly showed that the 0.5% APP diet significantly up-regulates slower myosin-heavy-chain (MyHC) isoform ratios (IIx and IIa relative to total MyHC) and myoglobin expression in lower hind-limb muscles examined (P < 0.05). There was a trend to increased fatigue resistance detected from measurements of relative isometric plantar-flexion force torque generated by a stimulus train delivered to the tibial nerve (F(98, 1372) = 1.246, P = 0.0574). Importantly, there was no significant difference in the animal body-phenotypes or locomotor activity shown as total moving distance in light and dark periods. Therefore, the present study encourages the notion that even low APP-intake may increase the proportions of fatigue-resistant myofibers, and has promise as a strategy for modifying performance in human sports and improving function in age-related muscle atrophy.
Collapse
Affiliation(s)
- Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Shinpei Okamoto
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Hideo Miyahara
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Mariko Akahoshi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan.,Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mai-Khoi Q Do
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Hideaki Ohtsubo
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Komiya
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Mulan Qahar
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Toshiaki Waga
- Fundamental Research Laboratory, Asahi Breweries, Ltd., Moriya, Ibaraki, Japan.,Wakodo, Ltd., Chofu, Tokyo, Japan
| | - Koichi Nakazato
- Department of Exercise Physiology, Graduate School of Health and Sport Sciences, Nippon Sport Science University, Tokyo, Japan
| | - Yoshihide Ikeuchi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Judy E Anderson
- Department of Biological Sciences, Faculty of Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
188
|
Viggiano E, Mollica MP, Lionetti L, Cavaliere G, Trinchese G, De Filippo C, Chieffi S, Gaita M, Barletta A, De Luca B, Crispino M, Monda M. Effects of an High-Fat Diet Enriched in Lard or in Fish Oil on the Hypothalamic Amp-Activated Protein Kinase and Inflammatory Mediators. Front Cell Neurosci 2016; 10:150. [PMID: 27375435 PMCID: PMC4899473 DOI: 10.3389/fncel.2016.00150] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/27/2016] [Indexed: 01/10/2023] Open
Abstract
The high fat diet (HFD) rich in lard induces obesity, inflammation and oxidative stress, and the deregulation of hypothalamic nuclei plays an important role in this mechanism. One important factor involved in the food intake and inflammation is adenosine monophosphate-dependent kinase (AMPK), a serine/threonine kinase activated by phosphorylation. Omega (ω)3-polyunsaturated fatty acids (PUFA) are dietary compounds known to attenuate the obesity-related diseases, although the molecular mechanisms underlying their actions in the hypothalamus are not completely understood. We hypothesized that the beneficial effects of PUFA may be mediated by AMPK in the hypothalamus. To this aim, rats were fed a control diet (CD), or isocaloric HFD containing either fish oil (FD; rich in ω3-PUFA) or lard for 6 weeks, and the activation of AMPK, inflammatory state (IKKβ, TNF-α) and oxidative stress were analyzed in the hypothalamus. In addition, we also studied serum lipid profile, homeostatic model assessment (HOMA) index, and pro-inflammatory parameters. Our results showed, at the hypothalamic level of LD-fed rats, an increase of AMPK activation, inflammation and oxidative stress, while no modifications were detected in FD-fed animals compared to CD. In addition body weight gain, serum lipid profile, pro-inflammatory parameters and insulin resistance were reduced in FD animals compared to LD. In conclusion, our data indicate that the substitution of saturated by unsaturated fatty acids in the diet has beneficial effects on modulation of hypothalamic inflammation and function in obesity, underlying, at hypothalamic level, the interaction among insulin and/or leptin resistance, AMPK activation and hyperphagia.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Experimental Medicine-Section of Human Physiology, Second University of NaplesNaples, Italy; Department of Medicine, University of PadovaPadua, Italy
| | | | - Lillà Lionetti
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II Naples, Italy
| | | | - Chiara De Filippo
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Sergio Chieffi
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| | - Marcello Gaita
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Antonio Barletta
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Bruno De Luca
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine-Section of Human Physiology, Second University of Naples Naples, Italy
| |
Collapse
|
189
|
Gopalan V, Michael N, Ishino S, Lee SS, Yang AY, Bhanu Prakash KN, Yaligar J, Sadananthan SA, Kaneko M, Zhou Z, Satomi Y, Hirayama M, Kamiguchi H, Zhu B, Horiguchi T, Nishimoto T, Velan SS. Effect of Exercise and Calorie Restriction on Tissue Acylcarnitines, Tissue Desaturase Indices, and Fat Accumulation in Diet-Induced Obese Rats. Sci Rep 2016; 6:26445. [PMID: 27197769 PMCID: PMC4873816 DOI: 10.1038/srep26445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/29/2016] [Indexed: 11/13/2022] Open
Abstract
Both exercise and calorie restriction interventions have been recommended for inducing weight-loss in obese states. However, there is conflicting evidence on their relative benefits for metabolic health and insulin sensitivity. This study seeks to evaluate the differential effects of the two interventions on fat mobilization, fat metabolism, and insulin sensitivity in diet-induced obese animal models. After 4 months of ad libitum high fat diet feeding, 35 male Fischer F344 rats were grouped (n = 7 per cohort) into sedentary control (CON), exercise once a day (EX1), exercise twice a day (EX2), 15% calorie restriction (CR1) and 30% calorie restriction (CR2) cohorts. Interventions were carried out over a 4-week period. We found elevated hepatic and muscle long chain acylcarnitines with both exercise and calorie restriction, and a positive association between hepatic long chain acylcarnitines and insulin sensitivity in the pooled cohort. Our result suggests that long chain acylcarnitines may not indicate incomplete fat oxidation in weight loss interventions. Calorie restriction was found to be more effective than exercise in reducing body weight. Exercise, on the other hand, was more effective in reducing adipose depots and muscle triglycerides, favorably altering muscle/liver desaturase activity and improving insulin sensitivity.
Collapse
Affiliation(s)
- Venkatesh Gopalan
- Laboratory of Molecular Imaging, Singapore Bio Imaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore
| | | | - Swee Shean Lee
- Laboratory of Molecular Imaging, Singapore Bio Imaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore
| | | | - K N Bhanu Prakash
- Laboratory of Molecular Imaging, Singapore Bio Imaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore
| | - Jadegoud Yaligar
- Laboratory of Molecular Imaging, Singapore Bio Imaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore
| | | | | | | | | | | | - Bin Zhu
- Takeda Singapore Pte Ltd., Singapore
| | - Takashi Horiguchi
- Molecular Imaging Centre, National Institute of Radiological Sciences, Chiba, Japan
| | | | - S Sendhil Velan
- Laboratory of Molecular Imaging, Singapore Bio Imaging Consortium, Agency for Science Technology and Research (A*STAR), Singapore
| |
Collapse
|
190
|
Taivassalo T, Hussain SN. Contribution of the Mitochondria to Locomotor Muscle Dysfunction in Patients With COPD. Chest 2016; 149:1302-12. [DOI: 10.1016/j.chest.2015.11.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/09/2015] [Accepted: 11/24/2015] [Indexed: 11/29/2022] Open
|
191
|
Laurens C, Badin PM, Louche K, Mairal A, Tavernier G, Marette A, Tremblay A, Weisnagel SJ, Joanisse DR, Langin D, Bourlier V, Moro C. G0/G1 Switch Gene 2 controls adipose triglyceride lipase activity and lipid metabolism in skeletal muscle. Mol Metab 2016; 5:527-537. [PMID: 27408777 PMCID: PMC4921782 DOI: 10.1016/j.molmet.2016.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Recent data suggest that adipose triglyceride lipase (ATGL) plays a key role in providing energy substrate from triglyceride pools and that alterations of its expression/activity relate to metabolic disturbances in skeletal muscle. Yet little is known about its regulation. We here investigated the role of the protein G0/G1 Switch Gene 2 (G0S2), recently described as an inhibitor of ATGL in white adipose tissue, in the regulation of lipolysis and oxidative metabolism in skeletal muscle. METHODS We first examined G0S2 protein expression in relation to metabolic status and muscle characteristics in humans. We next overexpressed and knocked down G0S2 in human primary myotubes to assess its impact on ATGL activity, lipid turnover and oxidative metabolism, and further knocked down G0S2 in vivo in mouse skeletal muscle. RESULTS G0S2 protein is increased in skeletal muscle of endurance-trained individuals and correlates with markers of oxidative capacity and lipid content. Recombinant G0S2 protein inhibits ATGL activity by about 40% in lysates of mouse and human skeletal muscle. G0S2 overexpression augments (+49%, p < 0.05) while G0S2 knockdown strongly reduces (-68%, p < 0.001) triglyceride content in human primary myotubes and mouse skeletal muscle. We further show that G0S2 controls lipolysis and fatty acid oxidation in a strictly ATGL-dependent manner. These metabolic adaptations mediated by G0S2 are paralleled by concomitant changes in glucose metabolism through the modulation of Pyruvate Dehydrogenase Kinase 4 (PDK4) expression (5.4 fold, p < 0.001). Importantly, downregulation of G0S2 in vivo in mouse skeletal muscle recapitulates changes in lipid metabolism observed in vitro. CONCLUSION Collectively, these data indicate that G0S2 plays a key role in the regulation of skeletal muscle ATGL activity, lipid content and oxidative metabolism.
Collapse
Affiliation(s)
- Claire Laurens
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Pierre-Marie Badin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Katie Louche
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Aline Mairal
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Geneviève Tavernier
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - André Marette
- Department of Medicine, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | - Angelo Tremblay
- Department of Kinesiology, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | | | - Denis R Joanisse
- Department of Kinesiology, Canada; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Canada
| | - Dominique Langin
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Virginie Bourlier
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - Cedric Moro
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France.
| |
Collapse
|
192
|
Alternate-Day High-Fat Diet Induces an Increase in Mitochondrial Enzyme Activities and Protein Content in Rat Skeletal Muscle. Nutrients 2016; 8:203. [PMID: 27058555 PMCID: PMC4848672 DOI: 10.3390/nu8040203] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/25/2016] [Accepted: 03/30/2016] [Indexed: 11/20/2022] Open
Abstract
Long-term high-fat diet increases muscle mitochondrial enzyme activity and endurance performance. However, excessive calorie intake causes intra-abdominal fat accumulation and metabolic syndrome. The purpose of this study was to investigate the effect of an alternating day high-fat diet on muscle mitochondrial enzyme activities, protein content, and intra-abdominal fat mass in rats. Male Wistar rats were given a standard chow diet (CON), high-fat diet (HFD), or alternate-day high-fat diet (ALT) for 4 weeks. Rats in the ALT group were fed a high-fat diet and standard chow every other day for 4 weeks. After the dietary intervention, mitochondrial enzyme activities and protein content in skeletal muscle were measured. Although body weight did not differ among groups, the epididymal fat mass in the HFD group was higher than those of the CON and ALT groups. Citrate synthase and beta-hydroxyacyl CoA dehydrogenase activities in the plantaris muscle of rats in HFD and ALT were significantly higher than that in CON rats, whereas there was no difference between HFD and ALT groups. No significant difference was observed in muscle glycogen concentration or glucose transporter-4 protein content among the three groups. These results suggest that an alternate-day high-fat diet induces increases in mitochondrial enzyme activities and protein content in rat skeletal muscle without intra-abdominal fat accumulation.
Collapse
|
193
|
Makimura H, Stanley TL, Suresh C, De Sousa-Coelho AL, Frontera WR, Syu S, Braun LR, Looby SE, Feldpausch MN, Torriani M, Lee H, Patti ME, Grinspoon SK. Metabolic Effects of Long-Term Reduction in Free Fatty Acids With Acipimox in Obesity: A Randomized Trial. J Clin Endocrinol Metab 2016; 101:1123-33. [PMID: 26691888 PMCID: PMC4803166 DOI: 10.1210/jc.2015-3696] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT Increased circulating free fatty acids (FFAs) have been proposed to contribute to insulin resistance in obesity. Short-term studies have investigated the effects of acipimox, an inhibitor of hormone-sensitive lipase, on glucose homeostasis, but longer-term studies have not been performed. OBJECTIVE To test the hypothesis that long-term treatment with acipimox would reduce FFA and improve insulin sensitivity among nondiabetic, insulin-resistant, obese subjects. DESIGN, SETTING, PATIENTS, AND INTERVENTION At an academic medical center, 39 obese men and women were randomized to acipimox 250 mg thrice-daily vs identical placebo for 6 months. MAIN OUTCOME MEASURES Plasma lipids, insulin sensitivity, adiponectin, and mitochondrial function via assessment of the rate of post-exercise phosphocreatine recovery on (31)P-magnetic resonance spectroscopy as well as muscle mitochondrial density and relevant muscle gene expression. RESULTS Fasting glucose decreased significantly in acipimox-treated individuals (effect size, -6 mg/dL; P = .02), in parallel with trends for reduced fasting insulin (effect size, -6.8 μU/mL; P = .07) and HOMA-IR (effect size, -1.96; P = .06), and significantly increased adiponectin (effect size, +668 ng/mL; P = .02). Acipimox did not affect insulin-stimulated glucose uptake, as assessed by euglycemic, hyperinsulinemic clamp. Effects on muscle mitochondrial function and density and on relevant gene expression were not seen. CONCLUSION These data shed light on the long-term effects of FFA reduction on insulin sensitivity, other metabolic parameters, and muscle mitochondrial function in obesity. Reduced FFA achieved by acipimox improved fasting measures of glucose homeostasis, lipids, and adiponectin but had no effect on mitochondrial function, mitochondrial density, or muscle insulin sensitivity.
Collapse
Affiliation(s)
- Hideo Makimura
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Takara L Stanley
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Caroline Suresh
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Ana Luisa De Sousa-Coelho
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Walter R Frontera
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Stephanie Syu
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Laurie R Braun
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Sara E Looby
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Meghan N Feldpausch
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Martin Torriani
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Hang Lee
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Mary-Elizabeth Patti
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| | - Steven K Grinspoon
- Program in Nutritional Metabolism and Neuroendocrine Unit (H.M., T.L.S., C.S., S.S., L.R.B., S.E.L., M.N.F., S.K.G.), Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School (H.M., T.L.S., A.L.D.S.-C., L.R.B., S.E.L., M.T., H.L., M.-E.P., S.K.G.), Boston, Massachusetts 02115; Pediatric Endocrine Unit (T.L.S., L.R.B.), Massachusetts General Hospital, Boston, Massachusetts 02114; Research Division (A.L.D.S.-C., M.-E.P.), Joslin Diabetes Center, Boston, Massachusetts 02215; Department of Physical Medicine and Rehabilitation (W.R.F.), Vanderbilt University Medical Center, Nashville, Tennessee 37212; Department of Physical Medicine and Rehabilitation (W.R.F.), Harvard Medical School/Spaulding Rehabilitation Hospital, Boston, Massachusetts 02114; Department of Physiology (W.R.F.), University of Puerto Rico School of Medicine, San Juan, Puerto Rico 00936; Department of Radiology (M.T.), Massachusetts General Hospital, Boston, Massachusetts 02114; and MGH Biostatistics Center (H.L.), Massachusetts General Hospital and Harvard Medical Center, Boston, Massachusetts 02114
| |
Collapse
|
194
|
Zhou Y, Gu P, Shi W, Li J, Hao Q, Cao X, Lu Q, Zeng Y. MicroRNA-29a induces insulin resistance by targeting PPARδ in skeletal muscle cells. Int J Mol Med 2016; 37:931-8. [PMID: 26936652 PMCID: PMC4790643 DOI: 10.3892/ijmm.2016.2499] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 02/10/2016] [Indexed: 12/03/2022] Open
Abstract
Intrauterine growth retardation (IUGR) induces metabolic syndrome, which is often characterized by insulin resistance (IR), in adults. Previous research has shown that microRNAs (miRNAs or miRs) play a role in the target genes involved in this process, but the mechanisms remain unclear. In the present study, we examined miRNA profiles using samples of skeletal muscles from both IUGR and control rat offspring whose mothers were fed either a protein-restricted diet or a diet which involved normal amounts of protein during pregnancy, respectively. miR-29a was found to be upregulated in the skeletal muscles of IUGR offspring. The luciferase reporter assay confirmed the direct interaction between miR-29a and peroxisome proliferator-activated receptor δ (PPARδ). Overexpression of miR-29a in the skeletal muscle cell line C2C12 suppressed the expression of its target gene PPARδ, which, in turn, influenced the expression of its coactivator, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). Thus, PPARδ/PGC-1α-dependent signals together reduced insulin-dependent glucose uptake and adenosine triphosphate (ATP) production. Overexpression of miR-29a also caused a decrease in levels of glucose transporter 4 (GLUT4), the most important glucose transporter in skeletal muscle, which partially induced a decrease insulin-dependent glucose uptake. These findings provide evidence for a novel micro-RNA-mediated mechanism of PPARδ regulation, and we also noted the IR-promoting actions of miR-29a in skeletal muscles of IUGR.
Collapse
Affiliation(s)
- Yuehua Zhou
- Department of Obstetrics and Gynecology of Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200036, P.R. China
| | - Pingqing Gu
- Department of Clinical Laboratory, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weijie Shi
- Department of Obstetrics and Gynecology of Xinghua People's Hospital, Xinghua, Jiangsu 225700, P.R. China
| | - Jingyun Li
- Department of Clinical Laboratory, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qun Hao
- Department of Obstetrics and Gynecology, Nanjing General Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Xiaomei Cao
- Duman High School, Singapore 436895, Republic of Singapore
| | - Qin Lu
- Department of Obstetrics and Gynecology of Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200036, P.R. China
| | - Yu Zeng
- Department of Clinical Laboratory, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
195
|
Yamazaki M, Munetsuna E, Yamada H, Ando Y, Mizuno G, Murase Y, Kondo K, Ishikawa H, Teradaira R, Suzuki K, Ohashi K. Fructose consumption induces hypomethylation of hepatic mitochondrial DNA in rats. Life Sci 2016; 149:146-52. [PMID: 26869391 DOI: 10.1016/j.lfs.2016.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/11/2015] [Accepted: 02/06/2016] [Indexed: 10/22/2022]
Abstract
AIMS Fructose may play a crucial role in the pathogenesis of metabolic syndrome (MetS). However, the pathogenic mechanism of the fructose-induced MetS has not yet been investigated fully. Recently, several reports have investigated the association between mitochondrial DNA (mtDNA) and MetS. We examined the effect of fructose-rich diets on mtDNA content, transcription, and epigenetic changes. MAIN METHODS Four-week-old male Sprague-Dawley rats were offered a 20% fructose solution for 14weeks. We quantified mRNAs for hepatic mitochondrial genes and analyzed the mtDNA methylation (5-mC and 5-hmC) levels using ELISA kits. KEY FINDINGS Histological analysis revealed non-alcoholic fatty liver disease (NAFLD) in fructose-fed rats. Hepatic mtDNA content and transcription were higher in fructose-fed rats than in the control group. Global hypomethylation of mtDNA was also observed in fructose-fed rats. SIGNIFICANCE We showed that fructose consumption stimulates hepatic mtDNA-encoded gene expression. This phenomenon might be due to epigenetic changes in mtDNA. Fructose-induced mitochondrial epigenetic changes appear to be a novel mechanism underlying the pathology of MetS and NAFLD.
Collapse
Affiliation(s)
- Mirai Yamazaki
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake, Japan.
| | - Yoshitaka Ando
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Genki Mizuno
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Yuri Murase
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Kanako Kondo
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Hiroaki Ishikawa
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Ryoji Teradaira
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Koji Suzuki
- Department of Public Health, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Koji Ohashi
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan.
| |
Collapse
|
196
|
Lalia AZ, Dasari S, Johnson ML, Robinson MM, Konopka AR, Distelmaier K, Port JD, Glavin MT, Esponda RR, Nair KS, Lanza IR. Predictors of Whole-Body Insulin Sensitivity Across Ages and Adiposity in Adult Humans. J Clin Endocrinol Metab 2016; 101:626-34. [PMID: 26709968 PMCID: PMC4880121 DOI: 10.1210/jc.2015-2892] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CONTEXT Numerous factors are purported to influence insulin sensitivity including age, adiposity, mitochondrial function, and physical fitness. Univariate associations cannot address the complexity of insulin resistance or the interrelationship among potential determinants. OBJECTIVE The objective of the study was to identify significant independent predictors of insulin sensitivity across a range of age and adiposity in humans. DESIGN, SETTING, AND PARTICIPANTS Peripheral and hepatic insulin sensitivity were measured by two stage hyperinsulinemic-euglycemic clamps in 116 men and women (aged 19-78 y). Insulin-stimulated glucose disposal, the suppression of endogenous glucose production during hyperinsulinemia, and homeostatic model assessment of insulin resistance were tested for associations with 11 potential predictors. Abdominal subcutaneous fat, visceral fat (AFVISC), intrahepatic lipid, and intramyocellular lipid (IMCL) were quantified by magnetic resonance imaging and spectroscopy. Skeletal muscle mitochondrial respiratory capacity (state 3), coupling efficiency, and reactive oxygen species production were evaluated from muscle biopsies. Aerobic fitness was measured from whole-body maximum oxygen uptake (VO2 peak), and metabolic flexibility was determined using indirect calorimetry. RESULTS Multiple regression analysis revealed that AFVISC (P < .0001) and intrahepatic lipid (P = .002) were independent negative predictors of peripheral insulin sensitivity, whereas VO2 peak (P = .0007) and IMCL (P = .023) were positive predictors. Mitochondrial capacity and efficiency were not independent determinants of peripheral insulin sensitivity. The suppression of endogenous glucose production during hyperinsulinemia model of hepatic insulin sensitivity revealed percentage fat (P < .0001) and AFVISC (P = .001) as significant negative predictors. Modeling homeostatic model assessment of insulin resistance identified AFVISC (P < .0001), VO2 peak (P = .001), and IMCL (P = .01) as independent predictors. CONCLUSION The reduction in insulin sensitivity observed with aging is driven primarily by age-related changes in the content and distribution of adipose tissue and is independent of muscle mitochondrial function or chronological age.
Collapse
Affiliation(s)
- Antigoni Z Lalia
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Surendra Dasari
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Matthew L Johnson
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Matthew M Robinson
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Adam R Konopka
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Klaus Distelmaier
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - John D Port
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Maria T Glavin
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Raul Ruiz Esponda
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - K Sreekumaran Nair
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Ian R Lanza
- Divisions of Endocrinology and Metabolism (A.Z.L., M.L.J., M.M.R., A.R.K., K.D., R.R.E., K.S.N., I.R.L.), Biomedical Statistics and Informatics (S.D.), and Radiology (J.D.P., M.T.G.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|
197
|
Kakehi S, Tamura Y, Takeno K, Sakurai Y, Kawaguchi M, Watanabe T, Funayama T, Sato F, Ikeda SI, Kanazawa A, Fujitani Y, Kawamori R, Watada H. Increased intramyocellular lipid/impaired insulin sensitivity is associated with altered lipid metabolic genes in muscle of high responders to a high-fat diet. Am J Physiol Endocrinol Metab 2016; 310:E32-40. [PMID: 26487001 DOI: 10.1152/ajpendo.00220.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/16/2015] [Indexed: 01/07/2023]
Abstract
The accumulation of intramyocellular lipid (IMCL) is recognized as an important determinant of insulin resistance, and is increased by a high-fat diet (HFD). However, the effects of HFD on IMCL and insulin sensitivity are highly variable. The aim of this study was to identify the genes in muscle that are related to this inter-individual variation. Fifty healthy men were recruited for this study. Before and after HFD for 3 days, IMCL levels in the tibialis anterior were measured by (1)H magnetic resonance spectroscopy, and peripheral insulin sensitivity was evaluated by glucose infusion rate (GIR) during the euglycemic-hyperinsulinemic clamp. Subjects who showed a large increase in IMCL and a large decrease in GIR by HFD were classified as high responders (HRs), and subjects who showed a small increase in IMCL and a small decrease in GIR were classified as low responders (LRs). In five subjects from each group, the gene expression profile of the vastus lateralis muscle was analyzed by DNA microarray analysis. Before HFD, gene expression profiles related to lipid metabolism were comparable between the two groups. Gene Set Enrichment Analysis demonstrated that five gene sets related to lipid metabolism were upregulated by HFD in the HR group but not in the LR group. Changes in gene expression patterns were confirmed by qRT-PCR using more samples (LR, n = 9; HR, n = 11). These results suggest that IMCL accumulation/impaired insulin sensitivity after HFD is closely associated with changes in the expression of genes related to lipid metabolism in muscle.
Collapse
Affiliation(s)
- Saori Kakehi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshifumi Tamura
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan;
| | - Kageumi Takeno
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuko Sakurai
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Minako Kawaguchi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takahiro Watanabe
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Funayama
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumihiko Sato
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shin-Ichi Ikeda
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Kanazawa
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshio Fujitani
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryuzo Kawamori
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan; Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan; and Center for Molecular Diabetology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
198
|
Shoar Z, Goldenthal MJ, De Luca F, Suarez E. Mitochondrial DNA content and function, childhood obesity, and insulin resistance. Endocr Res 2016; 41:49-56. [PMID: 26513277 DOI: 10.3109/07435800.2015.1068797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The objectives of our study were to compare the mitochondrial enzyme activity between obese and non-obese children and to assess the association between mitochondrial DNA content and function and markers of metabolic syndrome. METHODS Clinical and anthropometric data of obese and normal-weight children ages 2-18 years were collected. We collected buccal swabs for mitochondrial respiratory enzymes (complex I, IV, and Citrate Synthase). In obese children only, serum levels of metabolic parameters and mitochondrial DNA from mononuclear cells were quantitated. RESULTS We recruited 75 obese and 65 normal-weight children. There was no difference in respiratory complex enzyme activity levels between obese and normal-weight subjects. In obese subjects, mitochondrial to nuclear DNA (mt/nDNA) ratio was significantly correlated with BMI Z-score and BMI percentile (p < 0.05, and p < 0.01, respectively), and the strength of this correlation was proportionate to the degree of obesity. We did not find any association between mt/nDNA ratio and metabolic parameters. We observed a significant positive association between complex IV activity and fasting insulin level (p < 0.05). Finally, fasting insulin explained 45% of the variation in the complex IV activity level (p < 0.05). CONCLUSION Our findings indicate that mitochondrial DNA content is directly related to obesity, but not to the markers of metabolic syndrome/insulin resistance in children. Longitudinal studies involving larger samples are needed to confirm our findings and help elucidate the relationship between mitochondrial function, adiposity, and insulin resistance.
Collapse
Affiliation(s)
| | - Michael J Goldenthal
- b Section of Child Neurology, St. Christopher's Hospital for Children, Drexel University College of Medicine , Philadelphia , PA , USA
| | | | | |
Collapse
|
199
|
Liao K, Yan J, Mai K, Ai Q. Dietary lipid concentration affects liver mitochondrial DNA copy number, gene expression and DNA methylation in large yellow croaker (Larimichthys crocea). Comp Biochem Physiol B Biochem Mol Biol 2015; 193:25-32. [PMID: 26692128 DOI: 10.1016/j.cbpb.2015.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 12/29/2022]
Abstract
In response to changes in energy demand and nutrient supply, the organism regulates mitochondrial metabolic status to coordinate ATP production. To survey mitochondrial metabolic adaptation in response to dietary lipid concentration, citrate synthase (EC 2.3.3.1, CS) activity, the expression of several mitochondrial transcription factors, mitochondrial DNA (mtDNA) copy number, mitochondrial gene expression, mtDNA methylation, and oxidative stress parameters were analyzed in the liver of large yellow croaker fed one of three diets with a low (6%), moderate (12%, the control diet) or high (18%) crude lipid content for 70 d. MtDNA copy number was significantly increased in the low- and high-lipid groups compared to the control. The transcription of cytochrome c oxidase 1 (COX1), COX2, COX3, ATP synthase 6 (ATPase 6), 12S rRNA and 16S rRNA was also significantly increased in the low-lipid group compared with the control, while the transcription of these genes in the high-lipid group was unchanged. Moreover, D-loop (displacement loop) methylation in the high-lipid group was significantly higher than the control. The increase in mtDNA copy number and mitochondrial transcription might be a compensatory mechanism that matches ATP supply to demand under a low-lipid diet, while the increase of mtDNA copy number with unchanged mitochondrial transcription in the high-lipid group probably came from the increase of D-loop methylation.
Collapse
Affiliation(s)
- Kai Liao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Jing Yan
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China.
| |
Collapse
|
200
|
Jeromson S, Gallagher IJ, Galloway SDR, Hamilton DL. Omega-3 Fatty Acids and Skeletal Muscle Health. Mar Drugs 2015; 13:6977-7004. [PMID: 26610527 PMCID: PMC4663562 DOI: 10.3390/md13116977] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle is a plastic tissue capable of adapting and mal-adapting to physical activity and diet. The response of skeletal muscle to adaptive stimuli, such as exercise, can be modified by the prior nutritional status of the muscle. The influence of nutrition on skeletal muscle has the potential to substantially impact physical function and whole body metabolism. Animal and cell based models show that omega-3 fatty acids, in particular those of marine origin, can influence skeletal muscle metabolism. Furthermore, recent human studies demonstrate that omega-3 fatty acids of marine origin can influence the exercise and nutritional response of skeletal muscle. These studies show that the prior omega-3 status influences not only the metabolic response of muscle to nutrition, but also the functional response to a period of exercise training. Omega-3 fatty acids of marine origin therefore have the potential to alter the trajectory of a number of human diseases including the physical decline associated with aging. We explore the potential molecular mechanisms by which omega-3 fatty acids may act in skeletal muscle, considering the n-3/n-6 ratio, inflammation and lipidomic remodelling as possible mechanisms of action. Finally, we suggest some avenues for further research to clarify how omega-3 fatty acids may be exerting their biological action in skeletal muscle.
Collapse
Affiliation(s)
- Stewart Jeromson
- Health and Exercise Sciences Research Group, School of Sport, University of Stirling, Stirling, FK9 4LA Scotland, UK.
| | - Iain J Gallagher
- Health and Exercise Sciences Research Group, School of Sport, University of Stirling, Stirling, FK9 4LA Scotland, UK.
| | - Stuart D R Galloway
- Health and Exercise Sciences Research Group, School of Sport, University of Stirling, Stirling, FK9 4LA Scotland, UK.
| | - D Lee Hamilton
- Health and Exercise Sciences Research Group, School of Sport, University of Stirling, Stirling, FK9 4LA Scotland, UK.
| |
Collapse
|